scholarly journals Antigenic Modulation of CD6 By Itolizumab Is a New Mechanism for Effector T Cell Inhibition

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 995-995
Author(s):  
Dalena Chu ◽  
Valeria Marrocco ◽  
Phoi Tiet ◽  
Jeanette Ampudia ◽  
Stephen Connelly ◽  
...  

Abstract Introduction Acute Graft-Versus-Host Disease (aGVHD) is a serious complication of hematopoietic stem cell transplantation (HSCT) that is primarily driven by alloreactive T cells. CD6 is a costimulatory receptor primarily expressed on T cells that promotes synapse formation, T cell activation, and migration into tissues by engaging with its ligand, activated leukocyte cell adhesion molecule (ALCAM). CD6 is expressed on reconstituting T cells soon after HSCT (Rambaldi et al., 2019) and early studies have shown that ex-vivo depletion of CD6 + donor cells prior to HSCT decreases the incidence of aGVHD (Soiffer et al., 1992; Soiffer et al., 1998). The reduced levels of aGVHD were attributed to an increased prevalence of CD6 - T cells that were less alloreactive (Rasmussen et al., 1994). Therefore, modulating activity of the CD6-ALCAM pathway may ameliorate aGVHD. Itolizumab is a humanized anti-CD6 monoclonal antibody that was previously described to block the engagement of ALCAM, thereby inhibiting T cell activity and trafficking. Here, we elucidate a second and highly novel mechanism in which antibody-mediated loss of CD6 from the surface of T cells results in CD6 low T cells that are hyporesponsive to T cell stimulation. Methods To assess the molecular mechanisms for itolizumab-induced loss of cell surface CD6, peripheral blood mononuclear cells (PBMCs) were exposed to different conditions including treatment with selected protease inhibitors, a membrane inhibitor, or Fc receptor blocking antibodies in the presence of itolizumab. Furthermore, B cells, NK cells, and monocytes were isolated from PBMCs and mixed with T cells in combination with itolizumab to evaluate cell contact requirements for loss. Following treatment, cell surface levels of CD6 protein were assessed by flow cytometry using a non-competing anti-CD6 detection antibody while analysis of full length CD6 was detected by western blot from total protein lysate. The soluble form of CD6 (sCD6) was analyzed by electrochemiluminescence and immunoprecipitation from supernatant of treated cells. Results Following treatment with itolizumab, the levels of cell surface CD6 was reduced as assessed by flow cytometry and western blot. Concurrent with a loss of cell surface CD6, levels of sCD6 in the cell supernatant increased, suggesting CD6 is primarily cleaved from the cell surface rather than internalized. Characterization of the cleaved sCD6 product by western blot revealed a 30KDa product. Itolizumab-induced changes in both levels of cell surface CD6 and sCD6 was abrogated by both 4-benzenesulfonyl fluoride hydrochloride (AEBSF), a serine protease inhibitor and cytochalasin D, an inhibitor of actin polymerization that prevents movement within the cell membrane and blocks endocytic trafficking. This suggests that a membrane-bound serine protease is responsible for cleavage. Itolizumab-induced CD6 cleavage did not occur with T cells alone and required the presence of other immune cells including monocytes and NK cells, but not B cells (Figure 1). In addition, when monocytes and T cells were separated by a membrane, cleavage of CD6 was not observed in the presence of itolizumab, indicating that cell-to-cell contact is required. Furthermore, blocking antibodies indicated that functional Fc receptors, especially FcγRIA, were required, suggesting that binding of itolizumab to both an Fc receptor on monocytes and CD6 on T cells predicated the cleavage event. The resulting CD6 low T cells were hyporesponsive to T cell stimulation, even in the absence of itolizumab, as indicated by reduced expression of PD-1, CD71 and CD25 as well as inflammatory cytokines (Figure 2). Inhibition was observed for both naïve and effector/memory T cell subsets. Conclusions Our results reveal a novel mechanism of antigenic modulation by itolizumab in which CD6 is cleaved from the T cell surface and released in a soluble form. Cleavage of CD6 occurs via a membrane-bound serine protease and appears dependent upon the engagement of itolizumab with Fc receptor(s) present on monocytes and NK cells. The loss of cell surface CD6 results in T cells with reduced responses to TCR-mediated stimulation. As such, treatment of aGVHD patients with itolizumab may reduce the alloreactivity of donor T cells, ameliorating disease symptoms and improving the clinical outcomes in these patients. Figure 1 Figure 1. Disclosures Chu: Equillium: Current Employment. Marrocco: Equillium, Inc.: Current Employment. Tiet: Equillium, Inc.: Current Employment. Ampudia: Equillium, Inc.: Current Employment. Connelly: Equillium: Current Employment, Divested equity in a private or publicly-traded company in the past 24 months, Membership on an entity's Board of Directors or advisory committees. Ng: Equillium: Current Employment, Current equity holder in publicly-traded company, Divested equity in a private or publicly-traded company in the past 24 months.

Blood ◽  
2005 ◽  
Vol 106 (3) ◽  
pp. 779-786 ◽  
Author(s):  
Kent S. Boles ◽  
Winfried Barchet ◽  
Tom Diacovo ◽  
Marina Cella ◽  
Marco Colonna

AbstractThe tumor suppressor in lung cancer-1 (TSLC1) gene is frequently silenced in human lung carcinomas, and its expression suppresses tumorigenesis in nude mice. TSLC1 encodes a cell-surface protein called Necl-2 that belongs to the Nectin and Nectin-like (Necl) family of molecules. Necl-2 mediates epithelial cell junctions by homotypic contacts and/or heterotypic interactions with other Nectins and Necls. Thus, it inhibits tumorigenesis by ensuring that epithelial cells grow in organized layers. Here, we demonstrate that natural killer (NK) cells and CD8+ T cells recognize Necl-2 through a receptor known as class I-restricted T-cell–associated molecule (CRTAM), which is expressed only on activated cells. CRTAM–Necl-2 interactions promote cytotoxicity of NK cells and interferon γ (IFN-γ) secretion of CD8+ T cells in vitro as well as NK cell–mediated rejection of tumors expressing Necl-2 in vivo. These results provide evidence for an additional mechanism of tumor suppression mediated by TSLC1 that involves cytotoxic lymphocytes. Furthermore, they reveal Necl-2 as one of the molecular targets that allows the immunosurveillance network to distinguish tumor cells from normal cells.


Blood ◽  
2011 ◽  
Vol 118 (18) ◽  
pp. 4902-4909 ◽  
Author(s):  
Fu Jun Li ◽  
Yoshiki Kubagawa ◽  
Matthew K. McCollum ◽  
Landon Wilson ◽  
Tomoko Motohashi ◽  
...  

Abstract The association of an IgM-Fc receptor (FcμR) with chronic lymphocytic leukemia (CLL) was suggested more than 30 years ago, but its authenticity has never been formally addressed. We examined the expression of the recently identified FcμR by B and T cells in CLL patients using receptor-specific monoclonal antibodies. CLL B cells (CD5+/CD19+) expressed much higher levels of FcμR on their cell surface than B cells from healthy donors. Such enhanced expression was more evident in immunoglobulin heavy chain variable region (IGHV)–mutated, CD38− or early Rai-stage CLL than in IGHV-unmutated, CD38+, or advanced Rai-stage CLL. Intriguingly, surface FcμR levels also were significantly elevated in the non-CLL B cells (CD5−/CD19+) and T cells (CD5+/CD19−), especially in IGHV-mutated CLL. CLL patients also had high serum titers of FcμR compared with healthy donors, and serum FcμR levels correlated significantly with circulating lymphocyte numbers but not with the IGHV mutation status or Rai stage. The serum FcμR was resolved as an ∼ 40-kDa protein, distinct from the cell surface FcμR of ∼ 60 kDa, and it was produced by both CLL B and non-CLL B cells. Mass spectrometric analysis revealed that the serum FcμR is a soluble form of the receptor encoded by an alternatively spliced FcμR transcript. These findings indicate enhanced levels of both membrane-bound and soluble forms of FcμR in CLL patients.


1991 ◽  
Vol 174 (6) ◽  
pp. 1407-1415 ◽  
Author(s):  
L L Spruyt ◽  
M J Glennie ◽  
A D Beyers ◽  
A F Williams

Crosslinking of CD2 antigen on T lymphocytes and natural killer (NK) cells leads to a rise in cytoplasmic-free Ca2+ concentration ([Ca2+]i). However, CD2 seems unlikely to interact directly with the second messenger pathways since signaling via CD2 is poor in T cells that lack the T cell receptor (TCR) and is absent in L cells or insect cells that express CD2. In contrast, NK cells that are also TCR- can be triggered via CD2, but it is unclear as to whether the CD16 Fc receptor (FcR) may facilitate this effect. The CD16 transmembrane molecule is expressed in a complex with the zeta homodimer or the zeta/gamma heterodimer and these dimers are also associated with the TCR complex. Thus, it seemed that zeta chains may provide the link between signaling on NK cells and T cells. This could be tested on TCR- cells since when CD16 is transfected into T cells it is expressed in a complex with TCR zeta homodimer or the zeta/gamma heterodimer. At first, potentiation of CD2 signaling was seen on TCR- Jurkat cells expressing CD16, but this was found to be dependent on trace levels (1%) of IgG in F(ab')2 antibody preparations. With pure F(ab')2, the effect was lost. Signaling on a rat NK cell line was also re-examined with F(ab')2 antibodies that had no IgG contamination, and again no signal transduction via CD2 was seen. We thus conclude that there is no clear evidence for potent signaling via CD2 on cells that lack a TCR complex and that TCR zeta chain expressed at the cell surface is not sufficient to potentiate signaling via CD2 as measured by an increase in [Ca2+]i.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2675-2675
Author(s):  
Arghya Ray ◽  
Melissa R Junttila ◽  
Ting DU ◽  
Dena Sutimantanapi ◽  
Xi Chen ◽  
...  

Abstract Introduction: Adenosine is an anti-inflammatory and immunosuppressive metabolite, that signals to diminish activation and proliferation of cytotoxic T-cells, impair activity of natural killer cells and CD4 + effector T-cells, and promote the expansion of immunosuppressive cell types. CD73, a cell surface ecto-5'-nucleotidase, is required to convert AMP to adenosine and is a major catalyst of adenosine generation in the tumor microenvironment. Overexpression of CD73 is observed in many tumors and correlates with unfavorable clinical outcome. Bone marrow (BM) aspirates from multiple myeloma (MM) patients have shown increased adenosine levels correspond with disease progression [Horenstein et al. Mol Med. 2016,22:694-704] In addition to the adenosine rich feature of MM, multiple cell types within the MM BM niche express the enzymes required for adenosine production from both NAD and ATP precursors, including CD38, CD203a, CD39 and CD73. Previously, we demonstrated that dysfunctional plasmacytoid dendritic cells (pDCs) predominantly found in the BM of MM patients contribute to MM cell growth, survival, and suppression of antitumor immunity [Chauhan et al, Cancer Cell 2009, 16:309-323; Ray et al, Leukemia 2015, 29:1441-1444]. We recently discovered that the interaction between pDCs and MM cells increased CD73 transcript and protein levels in both cell types, implicating a role for adenosine signaling via CD73 signaling axis in MM. Together, these MM disease features indicate that reducing the level of adenosine via inhibition of CD73 may represent a unique vulnerability and treatment strategy for MM. Methods: To understand the functional consequence of CD73 inhibition in MM, autologous ex vivo cell assays using freshly isolated BM aspirates from MM patients were used to detect changes in immune cell function and MM cell viability upon treatment with OP-5558, a potent and selective CD73 small molecule inhibitor which is an analog of the clinical candidate, ORIC-533. The majority of BM samples utilized were from patients with relapsed or refractory MM after at least three lines of therapy including immunomodulatory drugs, proteasome inhibitors, and anti-CD38 monoclonal antibodies, as well as a patient with relapsed MM post BCMA-CAR-T therapy. Results: In BM aspirates from MM patients with relapsed refractory MM, CD73 inhibition by OP-5558 triggered activation of MM pDCs, evidenced by increased expression of CD40/CD83/CD86 (1.2-1.5-fold, OP-5558-treated versus untreated; p < 0.05; n=3). This inhibition of CD73 reversed immunosuppression in MM BM. Specifically, CD73 inhibitor OP-5558 stimulated T-cell activation, associated with increased CD69 cell surface expression on CD3 + T-cells (CD69 MFI:20% increase, treated versus control; p = 0.0031; n = 3). Moreover, CD8 + T-cells from these co-cultures enhanced cytolytic activity against patient MM cells, significantly decreasing autologous MM cell viability (mean 42% decrease in viability; treated versus control; p=0.014; n=5). Of note, OP-5558 treatment did not directly affect viability of MM cells when treated in isolation, indicating that the observed decreased viability occurs via enhanced cytotoxic T-cell activity. Importantly, we show that OP-5558 triggered significant MM cell lysis even within autologous MM bone marrow mononuclear cell (BMNC) cultures, confirming that CD73 inhibition restores MM-specific cytolytic activity of autologous patient T-cells in the MM BM microenvironment. (mean 37% decrease in viability; treated versus control; p=0.009; n=3). Conclusions: This study therefore demonstrates that: 1. CD73-mediated adenosine activity suppresses the cytolytic activity of T-cells against tumor cells in the MM BM milieu; and 2. CD73 inhibition can overcome immune suppression and restore lysis of MM cells by autologous T-cells. A clinical trial of potent, selective, orally bioavailable CD73 inhibitor ORIC-533 will examine the utility of CD73 inhibition to improve outcome in patients with relapsed refractory MM. Disclosures Junttila: ORIC Pharmaceuticals: Current Employment. Sutimantanapi: ORIC Pharmaceuticals: Current Employment. Chen: ORIC Pharmaceuticals: Current Employment. Warne: ORIC Pharmaceuticals: Current Employment. Chang: ORIC Pharmaceuticals: Current Employment. Blank: ORIC Pharmaceuticals: Current Employment. Wu: ORIC Pharmaceuticals: Current Employment. Moore: ORIC Pharmaceuticals: Current Employment. Ndubaku: ORIC Pharmaceuticals: Current Employment. Zavorotinskaya: ORIC Pharmaceuticals: Current Employment. Nadeem: GSK: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Adaptive Biotechnologies: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees. Friedman: ORIC Pharmaceuticals: Current Employment. Chauhan: C4 Therapeutics: Current equity holder in publicly-traded company; Stemline Therapeutics, Inc: Consultancy. Anderson: Millenium-Takeda: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Sanofi-Aventis: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Scientific Founder of Oncopep and C4 Therapeutics: Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company; AstraZeneca: Membership on an entity's Board of Directors or advisory committees; Mana Therapeutics: Membership on an entity's Board of Directors or advisory committees.


Cancers ◽  
2021 ◽  
Vol 13 (23) ◽  
pp. 5947
Author(s):  
Guranda Chitadze ◽  
Ulrike Wehkamp ◽  
Ottmar Janssen ◽  
Monika Brüggemann ◽  
Marcus Lettau

CD26/Dipeptidylpeptidase 4 is a transmembrane serine protease that cleaves off N-terminal dipeptides. CD26/DPP4 is expressed on several immune cell types including T and NK cells, dendritic cells, and activated B cells. A catalytically active soluble form of CD26/DPP4 can be released from the plasma membrane. Given its wide array of substrates and interaction partners CD26/DPP4 has been implicated in numerous biological processes and effects can be dependent or independent of its enzymatic activity and are exerted by the transmembrane protein and/or the soluble form. CD26/DPP4 has been implicated in the modulation of T-cell activation and proliferation and CD26/DPP4-positive T cells are characterized by remarkable anti-tumor properties rendering them interesting candidates for T cell-based immunotherapies. Moreover, especially in cutaneous T-cell lymphoma CD26/DPP4 expression patterns emerged as an established marker for diagnosis and treatment monitoring. Surprisingly, besides a profound knowledge on substrates, interaction partners, and associated signal transduction pathways, the precise role of CD26/DPP4 for T cell-based immune responses is only partially understood.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1185-1185
Author(s):  
Patrick P Ng ◽  
Wade Aaron ◽  
Evan Callihan ◽  
Golzar Hemmati ◽  
Che-Leung Law ◽  
...  

Abstract Introduction B-cell maturation antigen (BCMA) is a cell surface receptor highly and selectively expressed on normal plasma cells and transformed plasma cells in multiple myeloma (MM) patients. Upon ligand binding, BCMA initiates signals that promote the survival of MM cells and the production of immunosuppressive factors. Therapeutics that target BCMA are being investigated in the clinic, with encouraging preliminary results. HPN217 is a Tri-specific T Cell-Activating Construct (TriTAC) specific to BCMA, to serum albumin for half-life extension, and to CD3ε for redirecting T cells against MM cells. It is currently being evaluated in a phase 1 /2 clinical trial for relapsed or refractory MM (NCT04184050). Herein, we describe translational studies to examine factors that may impact the therapeutic efficacy of HPN217, including the target BCMA, in membrane-bound or soluble form, and concomitant or combination therapeutics such as γ-secretase inhibitor (GSI) and dexamethasone. Results To evaluate the effects of HPN217 against primary MM cells, we used a patient-derived 3D-culture system (3DTEBM) designed to recapitulate the biology within the bone marrow microenvironment. 3DTEBM seeded with bone marrow accessory cells and autologous plasma recreate niches along an oxygen gradient that enable the survival and expansion of autologous MM cells without additional nutrient supplements. 3DTEBM's were established from 5 MM patients with varying ratios of autologous CD3+ T cells to MM cells (0.15-0.6). Although the functional competence of the T cells was unknown, HPN217 was able to mediate MM cell killing in 80% of the cultures with up to 71% of MM cells eliminated at a T cell/MM cell ratio of 0.45. The anti-tumor efficacy of HPN217 correlated strongly (R 2 = 0.99) with BCMA expression on the MM cells as measured by flow cytometry, suggesting the number of target receptors can be a limiting factor in efficacy. Consistent with this result, pre-incubation of target cells with 1 or 10 μg/mL anti-BCMA reduced the activity of HPN217 in T cell-dependent cellular cytotoxicity (TDCC) assays using healthy donor T cells and MM cell lines. Soluble BCMA (sBCMA) is produced when the extracellular domain of BCMA is cleaved by γ-secretase. It may act as a sink for HPN217. There was no correlation between the activity of HPN217 and the quantity of sBCMA in 3DTEBM. However, in TDCC assays, the addition of 6.25, 25 and 100 nM recombinant BCMA respectively led to 4-, 9- and 28-fold increases in the EC 50 of HPN217. Taken together, these data underscore the importance of preserving BCMA on MM cells and reducing sBCMA in circulation. Interestingly, treatment of MM cell line RPMI8226 with the GSI LY-3039478 for 24 hours increased the cell surface expression of BCMA by 3.6 folds. Using RPMI8226 as target cells in the 3DTEBM system, LY-3039478 increased the killing efficacy of HPN217-redirected primary T cells by 1.9 folds. Dexamethasone (Dex) is used with other therapeutics for treating MM. It is also commonly given to manage cytokine release syndrome (CRS) caused by T cell engagers. We conducted TDCC assays in the presence of 0.07-300 nM Dex to simulate plasma concentrations relevant to dose levels of Dex premedication for CRS. The highest Dex concentrations caused ≤3-fold increases in the EC 50 of HPN217. Considering this and the plasma half-life of i.v. injected Dex at <5 h, the suppressive effect of Dex on the anti-tumor activity of HPN217-redirected T cells may be limited. We then evaluated if MM.1S-Luc cell line xenografts in NCG mice would be a suitable model to extend the above in vitro findings to an in vivo setting. Lesions in the spine, skull and femur in NCG mice treated with vehicle could be detected by bioluminescent imaging. All mice succumbed to the disease within 40 days. By contrast, animals treated with HPN217 were protected in a dose-dependent manner. Mice that received the highest dose remained 100% disease-free at the end of the study (Figure 1). Conclusions We demonstrated HPN217 mediated BCMA-dependent primary MM cell killing by autologous T cells, and that the density of BCMA target on the surface of MM cells and sBCMA affected the efficacy of HPN217 in cultures. GSI, which increased the expression of BCMA on MM cells, enhanced the efficacy of HPN217. On the other hand, Dex had limited negative effect. HPN217 in combination with approved and experimental MM therapeutics is being evaluated in the 3DTEBM and MM.1S-Luc models. Figure 1 Figure 1. Disclosures Ng: Harpoon Therapeutics: Current Employment, Current equity holder in publicly-traded company. Aaron: Harpoon Therapeutics: Current Employment, Current equity holder in publicly-traded company. Callihan: Harpoon Therapeutics: Current Employment, Current equity holder in publicly-traded company. Hemmati: Harpoon Therapeutics: Current Employment, Current equity holder in publicly-traded company. Law: Harpoon Therapeutics: Current Employment, Current equity holder in publicly-traded company. Azab: Cellatrix, LLC: Current Employment, Current holder of individual stocks in a privately-held company. Sun: Harpoon Therapeutics: Consultancy, Current equity holder in publicly-traded company, Ended employment in the past 24 months.


1983 ◽  
Vol 158 (3) ◽  
pp. 752-766 ◽  
Author(s):  
J Fujimoto ◽  
S Levy ◽  
R Levy

Sensitive enzyme-linked immunosorbent assays (ELISA) for the detection of human T cell antigens in soluble form have been developed. The assays use mouse monoclonal antibodies and specific anti-Leu sera prepared in rabbits by immunizing with Leu antigens absorbed to monoclonal antibody affinity columns. With these assays, Leu-1, -2, and -3 antigen signals from extracts of as few as 5 X 10(3) cells could be detected. When culture supernatants from various cell lines were tested, Leu-2 antigen, but not Leu-1 or Leu-3, was found to be present. Leu-2 antigen was present only in supernatants from T cell lines that expressed Leu-2 on their cell surface. Leu-2 antigen accumulated progressively in the supernatant of low density culture and its presence did not depend on cell proliferation or on fetal calf serum in the culture medium. The Leu-2 antigen in the supernatant was found to have only one Leu-2a determinant, whereas Leu-2 antigen from cell extracts had at least two determinants. The Leu-2 molecule was effectively purified from supernatant with an anti-Leu-2a affinity column. The purified Leu-2 antigen from supernatant of HPB-ALL cells was a single polypeptide chain of 27,000 mol wt, whereas Leu-2 antigen present on HPB-ALL cell surface was composed of two or more identical polypeptide chains of 33,000 mol wt linked by disulfide bonds. Normal human sera and sera from leukemia patients were also examined for the presence of the Leu-2 antigen. Normal human sera contained low levels of Leu-2 antigen but sera from Leu-2-positive leukemia patients had high levels. These results indicate that Leu-2 antigen is released from human T cells under physiological conditions.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 926-926
Author(s):  
Katrin M. Baltz ◽  
Matthias Krusch ◽  
Tina Baessler ◽  
Mercedes Kloss ◽  
Ingrid Kumbier ◽  
...  

Abstract Members of the TNF superfamily mediate multiple cellular functions, including cellular proliferation, differentiation and cell death. Many members of this protein family are shed from the cell surface as soluble forms, which affects cell-cell interactions by reduction of ligand densities and distally modulates effector cells bearing the respective receptor. The TNF family member Glucocorticoid-induced TNF Receptor (GITR) costimulates effector T cells, modulates apoptosis and NFkappaB and abrogates suppression of murine but not human regulatory T cells. Its cognate ligand GITRL has been found in various healthy tissues. Recently we reported that NK cells express GITR, while tumor cells express GITR ligand (GITRL), and GITR/GITRL interaction downregulates NK cell-mediated anti-tumor effector mechanisms like cytotoxicity and IFN-gamma production. Here we report that human tumor cells spontaneously release a soluble form of GITRL (sGITRL) detectable in culture supernatants by ELISA. Furthermore, we found elevated levels of sGITRL in sera of patients with various malignancies compared to healthy controls. We demonstrate that the release of GITRL from tumor cells can be blocked by inhibition of metalloproteinases, concomitantly causing accumulation of GITRL on the tumor cell surface as determined by FACS analysis. Upregulated GITRL surface expression further increased inhibition of NK cell anti-tumor effector mechanisms, while, in contrast, presence of sGITRL in cocultures of GITRL-expressing tumor cells and GITR-positive NK cells enhanced NK cell cytotoxicity and IFN-gamma production. Thus, in line with the results obtained with other TNF family members, conversion of membrane bound GITRL to its soluble form is associated with downregulation of its function, potentially due to blocking its cognate receptor. Thus, release of sGITRL substantially influences the interaction of tumor cells with NK cells. In addition, determination of sGITRL levels may be implemented as a diagnostic marker in patients with malignancies. Further prospective studies are currently being conducted addressing the value of GITRL as a tumor marker in different tumor entities.


2019 ◽  
Vol 5 (3) ◽  
pp. 63
Author(s):  
Alice Bayiyana ◽  
Samuel Okurut ◽  
Rose Nabatanzi ◽  
Godfrey Zziwa ◽  
David R. Boulware ◽  
...  

Despite improvement in the prognosis of HIV/AIDS (human immunodeficiency virus/acquired immune deficiency syndrome) patients on antiretroviral therapy (ART), cryptococcal meningitis (CM) still causes 10–15% mortality among HIV-infected patients. The immunological impact of ART on the CD4+ and CD8+ T cell repertoire during cryptococcal co-infection is unclear. We determined longitudinal phenotypic changes in T cell subsets among patients with CM after they initiated ART. We hypothesized that ART alters the clonotypic phenotype and structural composition of CD4+ and CD8+ T cells during CM co-infection. For this substudy, peripheral blood mononuclear cells (PBMC) were isolated at four time points from CM patients following ART initiation during the parent study (ClinicalTrials.gov number, NCT01075152). Phenotypic characterization of CD4+ and CD8+ T cells was done using T cell surface marker monoclonal antibodies by flow cytometry. There was variation in the expression of immunophenotypic markers defining central memory (CD27+CD45R0+), effector memory (CD45R0+CD27–), immune activation (CD38+ and Human Leucocyte Antigen DR (HLA-DR+), and exhaustion (Programmed cell death protein one (PD-1) in the CD4+ T cell subset. In comparison to the CD4+ T cell population, the CD8+ central memory subset declined gradually with minimal increase in the effector memory subset. Both CD4+ and CD8+ T cell immune exhaustion and activation markers remained elevated over 12 weeks. The relative surge and decline in the expression of T cell surface markers outlines a variation in the differentiation of CD4+ T cells during ART treatment during CM co-infection.


Author(s):  
Shannon L. McArdel ◽  
Anne-Sophie Dugast ◽  
Maegan E. Hoover ◽  
Arjun Bollampalli ◽  
Enping Hong ◽  
...  

AbstractRecombinant agonists that activate co-stimulatory and cytokine receptors have shown limited clinical anticancer utility, potentially due to narrow therapeutic windows, the need for coordinated activation of co-stimulatory and cytokine pathways and the failure of agonistic antibodies to recapitulate signaling by endogenous ligands. RTX-240 is a genetically engineered red blood cell expressing 4-1BBL and IL-15/IL-15Rα fusion (IL-15TP). RTX-240 is designed to potently and simultaneously stimulate the 4-1BB and IL-15 pathways, thereby activating and expanding T cells and NK cells, while potentially offering an improved safety profile through restricted biodistribution. We assessed the ability of RTX-240 to expand and activate T cells and NK cells and evaluated the in vivo efficacy, pharmacodynamics and tolerability using murine models. Treatment of PBMCs with RTX-240 induced T cell and NK cell activation and proliferation. In vivo studies using mRBC-240, a mouse surrogate for RTX-240, revealed biodistribution predominantly to the red pulp of the spleen, leading to CD8 + T cell and NK cell expansion. mRBC-240 was efficacious in a B16-F10 melanoma model and led to increased NK cell infiltration into the lungs. mRBC-240 significantly inhibited CT26 tumor growth, in association with an increase in tumor-infiltrating proliferating and cytotoxic CD8 + T cells. mRBC-240 was tolerated and showed no evidence of hepatic injury at the highest feasible dose, compared with a 4-1BB agonistic antibody. RTX-240 promotes T cell and NK cell activity in preclinical models and shows efficacy and an improved safety profile. Based on these data, RTX-240 is now being evaluated in a clinical trial.


Sign in / Sign up

Export Citation Format

Share Document